1
|
Wang T, Li MY, Pei Z, Chen QX, Cheng QS, Li Z. Down-regulation of platelet-derived growth factor receptor β in pericytes increases blood-brain barrier permeability and significantly enhances α-synuclein in a Parkinson's Disease 3D cell model in vitro under hyperglycemic condition. Tissue Cell 2025; 93:102751. [PMID: 39847894 DOI: 10.1016/j.tice.2025.102751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/24/2024] [Accepted: 01/15/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Parkinson's Disease (PD) often presents with a compromised blood-brain barrier (BBB), which hyperglycemia may exacerbate. Pericytes, a key cell for BBB integrity, are potential therapeutic targets for neurodegenerative disorders. Few studies have developed 3D PD cell models incorporating neurovascular units (NVU) through the co-culture of human endothelial, pericytes, astrocytes, and SH-SY5Y cells to evaluate BBB impairment and the role of pericytes under hyperglycemic condition. METHOD A 3D PD like cell model was developed using 6-OHDA-affected SH-SY5Y cells, combined with endothelial cells, pericytes, and astrocytes through the Real Architecture for Tissue (RAFT) 3D co-culture system. PD incorporating reduced (30 % and 89 %) PDGFRβ NVU (RPN) with or without hyperglycemic model (HM) were also established. BBB permeability to sodium fluorescein was assessed, and BBB impairment was evaluated using BBB-associated proteins (ZO-1, CD54, CD144), cell-specific proteins (CD31, GFAP, PDGFRβ, CD13), tyrosine hydroxylase (TH), α-synuclein, oligomeric α-synuclein, and α-synuclein (ser9). RESULTS PD 3D cell models incorporating RPN with or without hyperglycemia were successfully established in vitro. Graduately increased BBB impairment was observed in PD, PD with RPN, and PD with RPN combined with HM, indicated by decreased BBB-associated and cell-specific proteins, reduced TH, and increased α-synuclein, oligomeric α-synuclein, and α-synuclein (ser9) compared to the NVU model. CONCLUSION Reduced pericyte PDGFRβ could increase BBB permeability, accelerate PD progression, and exacerbate under hyperglycemic condition.
Collapse
Affiliation(s)
- Ting Wang
- Department of Neurology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong 510282, PR China.
| | - Meng-Yan Li
- Department of Neurology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong 510282, PR China.
| | - Zhong Pei
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road II, Guangzhou 510080, PR China.
| | - Qiu-Xia Chen
- Department of Neurology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong 510282, PR China; Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China.
| | - Qiu-Sheng Cheng
- Department of Neurology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong 510282, PR China.
| | - Ze Li
- Department of Neurology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong 510282, PR China.
| |
Collapse
|
2
|
Kirazli O, Ozkan M, Verimli U, Gulhan R, Arman A, Sehirli US. The effect of growth hormone on motor findings and dendrite morphology in an experimental Parkinson's disease model. Anat Sci Int 2025; 100:79-87. [PMID: 39085683 DOI: 10.1007/s12565-024-00790-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 07/17/2024] [Indexed: 08/02/2024]
Abstract
Approaches for the induction of neurogenesis and neuronal recovery through several modalities are gaining popularity in Parkinson's disease (PD). Growth hormone (GH) seems to have a role in the reversal of neural function following brain injury as well as in normal brain development and function; therefore, the use of GH may represent a feasible strategy in the management of PD. This experimental study aimed to evaluate the effect of growth hormone on motor function and dendrite morphology in rats with 6-hydroxydopamine (6-OHDA)-induced PD model. Thirty-six Sprague Dawley rats were included and randomly allocated into one of the six study groups: two controls and four treatment groups that received daily subcutaneous growth hormone injections for 21 days, 1, 2, and 3 months. PD model was induced through unilateral 6-OHDA injection to the nigrostriatal pathway. The following assessments were made: apomorphine rotation test, stepping test, and tissue examinations for tyrosine hydroxylase and dendrite morphology. The apomorphine rotation test and the stepping test confirmed the presence of PD. These tests as well as dendritic spine density/number and length assessments showed improvement in PD findings over time with GH administration. Findings of this study suggest that GH administration may improve dendrite morphology and motor function in the PD model, which may translate into symptom relief and quality of life improvement in patients with PD. Such potential benefits should be tested in robust clinical studies.
Collapse
Affiliation(s)
- Ozlem Kirazli
- Department of Anatomy, Marmara University School of Medicine, Marmara Universitesi Tip Fakultesi Anatomi Anabilim Dali, Basibuyuk, Maltepe, 34854, Istanbul, Turkey.
| | - Mazhar Ozkan
- Department of Anatomy, Namik Kemal University School of Medicine, Tekirdaǧ, Turkey
| | - Ural Verimli
- Department of Anatomy, Marmara University School of Medicine, Marmara Universitesi Tip Fakultesi Anatomi Anabilim Dali, Basibuyuk, Maltepe, 34854, Istanbul, Turkey
| | - Rezzan Gulhan
- Department of Pharmacology, Marmara University School of Medicine, Istanbul, Turkey
| | - Ahmet Arman
- Department of Medical Genetics, Marmara University School of Medicine, Istanbul, Turkey
| | - Umit Suleyman Sehirli
- Department of Anatomy, Marmara University School of Medicine, Marmara Universitesi Tip Fakultesi Anatomi Anabilim Dali, Basibuyuk, Maltepe, 34854, Istanbul, Turkey
| |
Collapse
|
3
|
Pardridge WM. Treatment of Parkinson's disease with biologics that penetrate the blood-brain barrier via receptor-mediated transport. Front Aging Neurosci 2023; 15:1276376. [PMID: 38035276 PMCID: PMC10682952 DOI: 10.3389/fnagi.2023.1276376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/27/2023] [Indexed: 12/02/2023] Open
Abstract
Parkinson's disease (PD) is characterized by neurodegeneration of nigral-striatal neurons in parallel with the formation of intra-neuronal α-synuclein aggregates, and these processes are exacerbated by neuro-inflammation. All 3 components of PD pathology are potentially treatable with biologics. Neurotrophins, such as glial derived neurotrophic factor or erythropoietin, can promote neural repair. Therapeutic antibodies can lead to disaggregation of α-synuclein neuronal inclusions. Decoy receptors can block the activity of pro-inflammatory cytokines in brain. However, these biologic drugs do not cross the blood-brain barrier (BBB). Biologics can be made transportable through the BBB following the re-engineering of the biologic as an IgG fusion protein, where the IgG domain targets an endogenous receptor-mediated transcytosis (RMT) system within the BBB, such as the insulin receptor or transferrin receptor. The receptor-specific antibody domain of the fusion protein acts as a molecular Trojan horse to ferry the biologic into brain via the BBB RMT pathway. This review describes the re-engineering of all 3 classes of biologics (neurotrophins, decoy receptor, therapeutic antibodies) for BBB delivery and treatment of PD. Targeting the RMT pathway at the BBB also enables non-viral gene therapy of PD using lipid nanoparticles (LNP) encapsulated with plasmid DNA encoding therapeutic genes. The surface of the lipid nanoparticle is conjugated with a receptor-specific IgG that triggers RMT of the LNP across the BBB in vivo.
Collapse
|
4
|
Firouzan B, Iravanpour F, Abbaszadeh F, Akparov V, Zaringhalam J, Ghasemi R, Maghsoudi N. Dipeptide mimetic of BDNF ameliorates motor dysfunction and striatal apoptosis in 6-OHDA-induced Parkinson's rat model: Considering Akt and MAPKs signaling. Behav Brain Res 2023; 452:114585. [PMID: 37467964 DOI: 10.1016/j.bbr.2023.114585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/21/2023]
Abstract
Parkinson's disease (PD) is a progressive and debilitating neurodegenerative disorder associated with motor and non-motor complaints. Dysregulation of neurotrophic factors and related signaling cascades have been reported to be common events in PD which is accompanied by dopaminergic (DA) neuron demise. However, the restoration of neurotrophic factors has several limitations. Bis-(N-monosuccinyl-L-methionyl-L-serine) heptamethylenediamide (BHME) is a dipeptide mimetic of brain-derived neurotrophic factor (BDNF) with reported anti-oxidant and neuroprotective effects in several experimental models. The current study has investigated the effect of BHME on 6-hydroxydopamine (6-OHDA)-caused motor anomalies in Wistar rats. In this regard, rats were treated daily with BHME (0.1 or 1 mg/kg) 1 h after 6-OHDA-caused damage until the twelfth day. Afterwards, motor behavior and DA neuron survival were evaluated via behavioral tests and immunohistochemistry (IHC) staining, respectively. Moreover, the activity of Akt, mitogen-activated protein kinases (MAPKs) family, and Bax/Bcl-2 ratio were evaluated by Western blotting. Our results indicated that BHME prevents motor dysfunction and DA cell death following 6-OHDA injection, and this improvement was in parallel with an enhancement in Akt activity, decrement of P38 phosphorylation, along with a reduction in Bax/Bcl-2 ratio. In conclusion, our findings indicated that BHME, as a mimetic of BDNF, can be considered for further research and is a promising therapeutic agent for PD therapy.
Collapse
Affiliation(s)
- Bita Firouzan
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farideh Iravanpour
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Abbaszadeh
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Valery Akparov
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jalal Zaringhalam
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Nader Maghsoudi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Kasanga EA, Han Y, Navarrete W, McManus R, Shifflet MK, Parry C, Barahona A, Manfredsson FP, Nejtek VA, Richardson JR, Salvatore MF. Differential expression of RET and GDNF family receptor, GFR-α1, between striatum and substantia nigra following nigrostriatal lesion: a case for diminished GDNF-signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530671. [PMID: 36909534 PMCID: PMC10002742 DOI: 10.1101/2023.03.01.530671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Although glial cell line-derived neurotrophic factor (GDNF) showed efficacy in preclinical and early clinical studies to alleviate parkinsonian signs in Parkinson's disease (PD), later trials did not meet primary endpoints, giving pause to consider further investigation. While GDNF dose and delivery methods may have contributed to diminished efficacy, one crucial aspect of these clinical studies is that GDNF treatment across all studies began ∼8 years after PD diagnosis; a time point representing several years after near 100% depletion of nigrostriatal dopamine markers in striatum and at least 50% in substantia nigra (SN), and is later than the timing of GDNF treatment in preclinical studies. With nigrostriatal terminal loss exceeding 70% at PD diagnosis, we utilized hemi-parkinsonian rats to determine if expression of GDNF family receptor, GFR-α1, and receptor tyrosine kinase, RET, differed between striatum and SN at 1 and 4 weeks following a 6-hydroxydopamine (6-OHDA) lesion. Whereas GDNF expression changed minimally, GFR-α1 expression decreased progressively in striatum and in tyrosine hydroxylase positive (TH+) cells in SN, correlating with reduced TH cell number. However, in nigral astrocytes, GFR-α1 expression increased. RET expression decreased maximally in striatum by 1 week, whereas in the SN, a transient bilateral increase occurred that returned to control levels by 4 weeks. Expression of brain-derived neurotrophic factor (BDNF) or its receptor, TrkB, were unchanged throughout lesion progression. Together, these results reveal that differential GFR-α1 and RET expression between the striatum and SN, and cell-specific differences in GFR-α1 expression in SN, occur during nigrostriatal neuron loss. Targeting loss of GDNF receptors appears critical to enhance GDNF therapeutic efficacy against nigrostriatal neuron loss. Significance Statement Although preclinical evidence supports that GDNF provides neuroprotection and improves locomotor function in preclinical studies, clinical data supporting its efficacy to alleviate motor impairment in Parkinson's disease patients remains uncertain. Using the established 6-OHDA hemi-parkinsonian rat model, we determined whether expression of its cognate receptors, GFR-α1 and RET, were differentially affected between striatum and substantia nigra in a timeline study. In striatum, there was early and significant loss of RET, but a gradual, progressive loss of GFR-α1. In contrast, RET transiently increased in lesioned substantia nigra, but GFR-α1 progressively decreased only in nigrostriatal neurons and correlated with TH cell loss. Our results indicate that direct availability of GFR-α1 may be a critical element that determines GDNF efficacy following striatal delivery. Highlights GDNF expression was minimally affected by nigrostriatal lesionGDNF family receptor, GFR-α1, progressively decreased in striatum and in TH neurons in SN.GFR-α1 expression decreased along with TH neurons as lesion progressedGFR-α1 increased bilaterally in GFAP+ cells suggesting an inherent response to offset TH neuron lossRET expression was severely reduced in striatum, whereas it increased in SN early after lesion induction.
Collapse
|
6
|
Paul G, Elabi OF. Microvascular Changes in Parkinson’s Disease- Focus on the Neurovascular Unit. Front Aging Neurosci 2022; 14:853372. [PMID: 35360216 PMCID: PMC8960855 DOI: 10.3389/fnagi.2022.853372] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/11/2022] [Indexed: 12/27/2022] Open
Abstract
Vascular alterations emerge as a common denominator for several neurodegenerative diseases. In Parkinson’s disease (PD), a number of observations have been made suggesting that the occurrence of vascular pathology is an important pathophysiological aspect of the disease. Specifically, pathological activation of pericytes, blood-brain barrier (BBB) disruption, pathological angiogenesis and vascular regression have been reported. This review summarizes the current evidence for the different vascular alterations in patients with PD and in animal models of PD. We suggest a possible sequence of vascular pathology in PD ranging from early pericyte activation and BBB leakage to an attempt for compensatory angiogenesis and finally vascular rarefication. We highlight different pathogenetic mechanisms that play a role in these vascular alterations including perivascular inflammation and concomitant metabolic disease. Awareness of the contribution of vascular events to the pathogenesis of PD may allow the identification of targets to modulate those mechanisms. In particular the BBB has for decades only been viewed as an obstacle for drug delivery, however, preservation of its integrity and/or modulation of the signaling at this interface between the blood and the brain may prove to be a new avenue to take in order to develop disease-modifying strategies for neurodegenerative disorders.
Collapse
Affiliation(s)
- Gesine Paul
- Translational Neurology Group, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Neurology, Scania University Hospital, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
- *Correspondence: Gesine Paul,
| | - Osama F. Elabi
- Translational Neurology Group, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
7
|
Eleftheriadou D, Evans RE, Atkinson E, Abdalla A, Gavins FKH, Boyd AS, Williams GR, Knowles JC, Roberton VH, Phillips JB. An alginate-based encapsulation system for delivery of therapeutic cells to the CNS. RSC Adv 2022; 12:4005-4015. [PMID: 35425456 PMCID: PMC8981497 DOI: 10.1039/d1ra08563h] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/22/2022] [Indexed: 12/21/2022] Open
Abstract
Treatment options for neurodegenerative conditions such as Parkinson's disease have included the delivery of cells which release dopamine or neurotrophic factors to the brain. Here, we report the development of a novel approach for protecting cells after implantation into the central nervous system (CNS), by developing dual-layer alginate beads that encapsulate therapeutic cells and release an immunomodulatory compound in a sustained manner. An optimal alginate formulation was selected with a view to providing a sustained physical barrier between engrafted cells and host tissue, enabling exchange of small molecules while blocking components of the host immune response. In addition, a potent immunosuppressant, FK506, was incorporated into the outer layer of alginate beads using electrosprayed poly-ε-caprolactone core–shell nanoparticles with prolonged release profiles. The stiffness, porosity, stability and ability of the alginate beads to support and protect encapsulated SH-SY5Y cells was demonstrated, and the release profile of FK506 and its effect on T-cell proliferation in vitro was characterized. Collectively, our results indicate this multi-layer encapsulation technology has the potential to be suitable for use in CNS cell delivery, to protect implanted cells from host immune responses whilst providing permeability to nutrients and released therapeutic molecules. Novel composite cell encapsulation system: dual-layer, micro-scale beads maintain cell survival while releasing immunomodulatory FK506 in a sustained manner. This biotechnology platform could be applicable for treatment of CNS and other disorders.![]()
Collapse
Affiliation(s)
- Despoina Eleftheriadou
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Rachael E Evans
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Emily Atkinson
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Ahmed Abdalla
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Francesca K H Gavins
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Ashleigh S Boyd
- UCL Institute of Immunity and Transplantation, Royal Free Hospital London UK
| | - Gareth R Williams
- UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - Jonathan C Knowles
- Biomaterials & Tissue Engineering, UCL Eastman Dental Institute London UK
| | - Victoria H Roberton
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| | - James B Phillips
- UCL Centre for Nerve Engineering, University College London London UK.,UCL School of Pharmacy, University College London London WC1N 1AX UK
| |
Collapse
|
8
|
Goulding SR, Anantha J, Collins LM, Sullivan AM, O'Keeffe GW. Growth differentiation factor 5: a neurotrophic factor with neuroprotective potential in Parkinson's disease. Neural Regen Res 2022; 17:38-44. [PMID: 34100424 PMCID: PMC8451580 DOI: 10.4103/1673-5374.314290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/08/2021] [Accepted: 01/24/2021] [Indexed: 11/04/2022] Open
Abstract
Parkinson's disease is the most common movement disorder worldwide, affecting over 6 million people. It is an age-related disease, occurring in 1% of people over the age of 60, and 3% of the population over 80 years. The disease is characterized by the progressive loss of midbrain dopaminergic neurons from the substantia nigra, and their axons, which innervate the striatum, resulting in the characteristic motor and non-motor symptoms of Parkinson's disease. This is paralleled by the intracellular accumulation of α-synuclein in several regions of the nervous system. Current therapies are solely symptomatic and do not stop or slow disease progression. One promising disease-modifying strategy to arrest the loss of dopaminergic neurons is the targeted delivery of neurotrophic factors to the substantia nigra or striatum, to protect the remaining dopaminergic neurons of the nigrostriatal pathway. However, clinical trials of two well-established neurotrophic factors, glial cell line-derived neurotrophic factor and neurturin, have failed to meet their primary end-points. This failure is thought to be at least partly due to the downregulation by α-synuclein of Ret, the common co-receptor of glial cell line-derived neurorophic factor and neurturin. Growth/differentiation factor 5 is a member of the bone morphogenetic protein family of neurotrophic factors, that signals through the Ret-independent canonical Smad signaling pathway. Here, we review the evidence for the neurotrophic potential of growth/differentiation factor 5 in in vitro and in vivo models of Parkinson's disease. We discuss new work on growth/differentiation factor 5's mechanisms of action, as well as data showing that viral delivery of growth/differentiation factor 5 to the substantia nigra is neuroprotective in the α-synuclein rat model of Parkinson's disease. These data highlight the potential for growth/differentiation factor 5 as a disease-modifying therapy for Parkinson's disease.
Collapse
Affiliation(s)
- Susan R. Goulding
- Department of Anatomy and Neuroscience, and Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Jayanth Anantha
- Department of Anatomy and Neuroscience, and Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Louise M. Collins
- Department of Anatomy and Neuroscience, and Cork Neuroscience Centre, University College Cork, Cork, Ireland
- Department of Physiology, University College Cork, Cork, Ireland
| | - Aideen M. Sullivan
- Department of Anatomy and Neuroscience, and Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Gerard W. O'Keeffe
- Department of Anatomy and Neuroscience, and Cork Neuroscience Centre, University College Cork, Cork, Ireland
| |
Collapse
|
9
|
Ferreira AFF, Binda KH, Real CC. The effects of treadmill exercise in animal models of Parkinson's disease: A systematic review. Neurosci Biobehav Rev 2021; 131:1056-1075. [PMID: 34688727 DOI: 10.1016/j.neubiorev.2021.10.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/04/2021] [Accepted: 10/18/2021] [Indexed: 12/18/2022]
Abstract
Parkinson's disease (PD) is a progressive disabling brain disorder. Physical exercise has been shown to alleviate the symptoms of PD and, consequently, improve patient quality of life. Exercise mechanisms involved in beneficial effects on PD have been widely investigated. This study aims to systematically review the literature on the use of treadmill exercise in PD animal models. The study was conducted according to Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA). Searches were conducted in MEDLINE, EMBASE, and ISI databases. In total, 78 studies were included. The dopaminergic system, behavior, neuroplasticity, neuroinflammation, mitochondria, and musculoskeletal systems were some of the outcomes evaluated by the selected studies. Based on the systematic review center for laboratory animal experimentation (SYRCLE) RoB tool, the methodologies revealed a high risk of bias and lack of information about study design, which needs attention for data reproducibility. This review can guide future studies that aim to fill existing gaps regarding the effects of treadmill exercise in PD animal models.
Collapse
Affiliation(s)
- Ana Flávia F Ferreira
- Laboratory of Cellular Neurobiology, Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Karina Henrique Binda
- Laboratory of Functional Neuroanatomy of Pain, Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil; Translational Neuropsychiatry Unit (TNU), Aarhus University, Aarhus, Denmark
| | - Caroline Cristiano Real
- Translational Neuropsychiatry Unit (TNU), Aarhus University, Aarhus, Denmark; Faculdade de Medicina (FMUSP), Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
10
|
Chen H, Teng Y, Chen X, Liu Z, Geng F, Liu Y, Jiang H, Wang Z, Yang L. Platelet-derived growth factor (PDGF)-BB protects dopaminergic neurons via activation of Akt/ERK/CREB pathways to upregulate tyrosine hydroxylase. CNS Neurosci Ther 2021; 27:1300-1312. [PMID: 34346167 PMCID: PMC8504523 DOI: 10.1111/cns.13708] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/18/2021] [Accepted: 06/29/2021] [Indexed: 12/18/2022] Open
Abstract
Aims The neurotropic growth factor PDGF‐BB was shown to have vital neurorestorative functions in various animal models of Parkinson's disease (PD). Previous studies indicated that the regenerative property of PDGF‐BB contributes to the increased intensity of tyrosine hydroxylase (TH) fibers in vivo. However, whether PDGF‐BB directly modulates the expression of TH, and the underlying mechanism is still unknown. We will carefully examine this in our current study. Method MPTP‐lesion mice received PDGF‐BB treatment via intracerebroventricular (i.c.v) administration, and the expression of TH in different brain regions was assessed by RT‐PCR, Western blot, and immunohistochemistry staining. The molecular mechanisms of PDGF‐BB‐mediated TH upregulation were examined by RT‐PCR, Western blot, ChIP assay, luciferase reporter assay, and immunocytochemistry. Results We validated a reversal expression of TH in MPTP‐lesion mice upon i.c.v administration of PDGF‐BB for seven days. Similar effects of PDGF‐BB‐mediated TH upregulation were also observed in MPP+‐treated primary neuronal culture and dopaminergic neuronal cell line SH‐SY5Y cells. We next demonstrated that PDGF‐BB rapidly activated the pro‐survival PI3K/Akt and MAPK/ERK signaling pathways, as well as the downstream CREB in SH‐SY5Y cells. We further confirmed the significant induction of p‐CREB in PDGF‐BB‐treated animals in vivo. Using a genetic approach, we demonstrated that the transcription factor CREB is critical for PDGF‐BB‐mediated TH expression. The activation and nucleus translocation of CREB were promoted in PDGF‐BB‐treated SH‐SY5Y cells, and the enrichment of CREB on the promoter region of TH gene was also increased upon PDGF‐BB treatment. Conclusion Our data demonstrated that PDGF‐BB directly regulated the expression of TH via activating the downstream Akt/ERK/CREB signaling pathways. Our finding will further support the therapeutic potential of PDGF‐BB in PD, and provide the possibility that targeting PDGF signaling can be harnessed as an adjunctive therapy in PD in the future.
Collapse
Affiliation(s)
- Huan Chen
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yan Teng
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xingmin Chen
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zhihao Liu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Fan Geng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yanzhuo Liu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Haisong Jiang
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ziyan Wang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Lu Yang
- Institute of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.,School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
11
|
Goulding SR, Lévesque M, Sullivan AM, Collins LM, O'Keeffe GW. Quinacrine and Niclosamide Promote Neurite Growth in Midbrain Dopaminergic Neurons Through the Canonical BMP-Smad Pathway and Protect Against Neurotoxin and α-Synuclein-Induced Neurodegeneration. Mol Neurobiol 2021; 58:3405-3416. [PMID: 33713017 DOI: 10.1007/s12035-021-02351-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/05/2021] [Indexed: 11/25/2022]
Abstract
Parkinson's disease is a neurodegenerative disorder characterised by nigrostriatal dopaminergic degeneration, and intracellular α-synuclein aggregation. Current pharmacological treatments are solely symptomatic so there is a need to identify agents that can slow or stop dopaminergic degeneration. One proposed class of therapeutics are neurotrophic factors which promote the survival of nigrostriatal dopaminergic neurons. However, neurotrophic factors need to be delivered directly to the brain. An alternative approach may be to identify pharmacological agents which can reach the brain to stimulate neurotrophic factor expression and/or their signalling pathways in dopaminergic neurons. BMP2 is a neurotrophic factor that is expressed in the human substantia nigra; exogenous BMP2 administration protects against dopaminergic degeneration in in vitro models of PD. In this study, we investigated the neurotrophic potential of two FDA-approved drugs, quinacrine and niclosamide, that are modulators of BMP2 signalling. We report that quinacrine and niclosamide, like BMP2, significantly increased neurite length, as a readout of neurotrophic action, in SH-SY5Y cells and dopaminergic neurons in primary cultures of rat ventral mesencephalon. We also show that these effects of quinacrine and niclosamide require the activation of BMP-Smad signalling. Finally, we demonstrate that quinacrine and niclosamide are neuroprotective against degeneration induced by the neurotoxins, MPP+ and 6-OHDA, and by viral-mediated overexpression of α-synuclein in vitro. Collectively, this study identifies two drugs, that are safe for use in patients' to 'are approved for human use, that exert neurotrophic effects on dopaminergic neurons through modulation of BMP-Smad signalling. This rationalises the further study of drugs that target the BMP-Smad pathway as potential neuroprotective pharmacotherapy for Parkinson's disease.
Collapse
Affiliation(s)
- Susan R Goulding
- Department of Biological Sciences, Munster Technological University, Cork, Ireland
- Department of Anatomy and Neuroscience and Cork Neuroscience Centre, University College Cork, Cork, Ireland
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Cervo Brain Research Centre, Université Laval, Quebec, QC, Canada
| | - Aideen M Sullivan
- Department of Anatomy and Neuroscience and Cork Neuroscience Centre, University College Cork, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Louise M Collins
- Department of Anatomy and Neuroscience and Cork Neuroscience Centre, University College Cork, Cork, Ireland.
- Department of Physiology, University College Cork, Cork, Ireland.
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience and Cork Neuroscience Centre, University College Cork, Cork, Ireland.
- APC Microbiome Institute, University College Cork, Cork, Ireland.
| |
Collapse
|
12
|
Goulding SR, Concannon RM, Morales-Prieto N, Villalobos-Manriquez F, Clarke G, Collins LM, Lévesque M, Wyatt SL, Sullivan AM, O'Keeffe GW. Growth differentiation factor 5 exerts neuroprotection in an α-synuclein rat model of Parkinson's disease. Brain 2021; 144:e14. [PMID: 33253375 DOI: 10.1093/brain/awaa367] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- Susan R Goulding
- Department of Anatomy and Neuroscience, University College Cork (UCC), Cork, Ireland.,Department of Biological Sciences, Cork Institute of Technology (CIT), Cork, Ireland
| | - Ruth M Concannon
- Department of Anatomy and Neuroscience, University College Cork (UCC), Cork, Ireland
| | - Noelia Morales-Prieto
- Department of Anatomy and Neuroscience, University College Cork (UCC), Cork, Ireland
| | | | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Sciences, UCC, Cork, Ireland.,APC Microbiome Ireland, UCC, Cork, Ireland.,Cork Neuroscience Centre, UCC, Cork, Ireland
| | - Louise M Collins
- Department of Anatomy and Neuroscience, University College Cork (UCC), Cork, Ireland
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Quebec, Canada
| | - Sean L Wyatt
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, UK
| | - Aideen M Sullivan
- Department of Anatomy and Neuroscience, University College Cork (UCC), Cork, Ireland.,APC Microbiome Ireland, UCC, Cork, Ireland.,Cork Neuroscience Centre, UCC, Cork, Ireland
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork (UCC), Cork, Ireland.,APC Microbiome Ireland, UCC, Cork, Ireland.,Cork Neuroscience Centre, UCC, Cork, Ireland
| |
Collapse
|
13
|
Miller KM, Mercado NM, Sortwell CE. Synucleinopathy-associated pathogenesis in Parkinson's disease and the potential for brain-derived neurotrophic factor. NPJ PARKINSONS DISEASE 2021; 7:35. [PMID: 33846345 PMCID: PMC8041900 DOI: 10.1038/s41531-021-00179-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
The lack of disease-modifying treatments for Parkinson’s disease (PD) is in part due to an incomplete understanding of the disease’s etiology. Alpha-synuclein (α-syn) has become a point of focus in PD due to its connection to both familial and idiopathic cases—specifically its localization to Lewy bodies (LBs), a pathological hallmark of PD. Within this review, we will present a comprehensive overview of the data linking synuclein-associated Lewy pathology with intracellular dysfunction. We first present the alterations in neuronal proteins and transcriptome associated with LBs in postmortem human PD tissue. We next compare these findings to those associated with LB-like inclusions initiated by in vitro exposure to α-syn preformed fibrils (PFFs) and highlight the profound and relatively unique reduction of brain-derived neurotrophic factor (BDNF) in this model. Finally, we discuss the multitude of ways in which BDNF offers the potential to exert disease-modifying effects on the basal ganglia. What remains unknown is the potential for BDNF to mitigate inclusion-associated dysfunction within the context of synucleinopathy. Collectively, this review reiterates the merit of using the PFF model as a tool to understand the physiological changes associated with LBs, while highlighting the neuroprotective potential of harnessing endogenous BDNF.
Collapse
Affiliation(s)
- Kathryn M Miller
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Neuroscience Graduate Program, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Natosha M Mercado
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Neuroscience Graduate Program, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA. .,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, USA.
| |
Collapse
|
14
|
Chu Y, Kordower JH. GDNF signaling in subjects with minimal motor deficits and Parkinson's disease. Neurobiol Dis 2021; 153:105298. [PMID: 33684514 DOI: 10.1016/j.nbd.2021.105298] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/18/2021] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
The failure of glial cell derived neurotropic factor to be efficacious in blinded clinical trials for Parkinson's disease may be due to alterations in signaling receptors and downstream signaling molecules. To test this hypothesis, brain sections were obtained from older adults with no motor deficit (n = 6), minimal motor deficits (n = 10), and clinical diagnosis of Parkinson's disease (n = 10) who underwent motor examination proximate to death. Quantitative unbiased stereology and densitometry were performed to analyze RET and phosphorylated ribosomal protein S6 expression in nigral neurons. Individuals with no motor deficit had extensive and intense RET and phosphorylated ribosomal protein S6 immunoreactive neurons in substantia nigra. The number and staining intensity of RET-immunoreactive neurons were reduced moderately in subjects with minimal motor deficits and severely reduced in Parkinson's disease relative to no motor deficit group. The number and staining intensity of phosphorylated ribosomal protein S6 was more markedly reduced in both subjects with minimal motor deficits and Parkinson's disease. Reductions in levels of RET and phosphorylated ribosomal protein S6 were recapitulated in a non-human primate genetic Parkinson's disease model based on over-expression of human mutant α-synuclein (A53T). These data indicate that for neurotrophic factors to be effective in patients with minimal motor deficits or PD, these factors would likely have to upregulate RET and phosphorylated ribosomal protein S6 immunoreactive neurons in substantia nigra .
Collapse
Affiliation(s)
- Yaping Chu
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Jeffrey H Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, United States of America.; ASU-Banner Neurodgenerative Disease Research Center, Arizona State University, Tempe, Arizona 85287, United States of America..
| |
Collapse
|
15
|
Virreira Winter S, Karayel O, Strauss MT, Padmanabhan S, Surface M, Merchant K, Alcalay RN, Mann M. Urinary proteome profiling for stratifying patients with familial Parkinson's disease. EMBO Mol Med 2021; 13:e13257. [PMID: 33481347 PMCID: PMC7933820 DOI: 10.15252/emmm.202013257] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/30/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
The prevalence of Parkinson's disease (PD) is increasing but the development of novel treatment strategies and therapeutics altering the course of the disease would benefit from specific, sensitive, and non-invasive biomarkers to detect PD early. Here, we describe a scalable and sensitive mass spectrometry (MS)-based proteomic workflow for urinary proteome profiling. Our workflow enabled the reproducible quantification of more than 2,000 proteins in more than 200 urine samples using minimal volumes from two independent patient cohorts. The urinary proteome was significantly different between PD patients and healthy controls, as well as between LRRK2 G2019S carriers and non-carriers in both cohorts. Interestingly, our data revealed lysosomal dysregulation in individuals with the LRRK2 G2019S mutation. When combined with machine learning, the urinary proteome data alone were sufficient to classify mutation status and disease manifestation in mutation carriers remarkably well, identifying VGF, ENPEP, and other PD-associated proteins as the most discriminating features. Taken together, our results validate urinary proteomics as a valuable strategy for biomarker discovery and patient stratification in PD.
Collapse
Affiliation(s)
- Sebastian Virreira Winter
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
- Present address:
OmicEra Diagnostics GmbHPlaneggGermany
| | - Ozge Karayel
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Maximilian T Strauss
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | | | | | | | - Roy N Alcalay
- Department of NeurologyColumbia UniversityNew YorkNYUSA
| | - Matthias Mann
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
- Novo Nordisk Foundation Center for Protein ResearchFaculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
16
|
Kambey PA, Kanwore K, Ayanlaja AA, Nadeem I, Du Y, Buberwa W, Liu W, Gao D. Failure of Glial Cell-Line Derived Neurotrophic Factor (GDNF) in Clinical Trials Orchestrated By Reduced NR4A2 (NURR1) Transcription Factor in Parkinson's Disease. A Systematic Review. Front Aging Neurosci 2021; 13:645583. [PMID: 33716718 PMCID: PMC7943926 DOI: 10.3389/fnagi.2021.645583] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/29/2021] [Indexed: 12/23/2022] Open
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative maladies with unforeseen complex pathologies. While this neurodegenerative disorder’s neuropathology is reasonably well known, its etiology remains a mystery, making it challenging to aim therapy. Glial cell-line derived neurotrophic factor (GDNF) remains an auspicious therapeutic molecule for treating PD. Neurotrophic factor derived from glial cell lines is effective in rodents and nonhuman primates, but clinical findings have been equivocal. Laborious exertions have been made over the past few decades to improve and assess GDNF in treating PD (clinical studies). Definitive clinical trials have, however, failed to demonstrate a survival advantage. Consequently, there seemed to be a doubt as to whether GDNF has merit in the potential treatment of PD. The purpose of this cutting edge review is to speculate as to why the clinical trials have failed to meet the primary endpoint. We introduce a hypothesis, “Failure of GDNF in clinical trials succumbed by nuclear receptor-related factor 1 (Nurr1) shortfall.” We demonstrate how Nurr1 binds to GDNF to induce dopaminergic neuron synthesis. Due to its undisputable neuro-protection aptitude, we display Nurr1 (also called Nr4a2) as a promising therapeutic target for PD.
Collapse
Affiliation(s)
- Piniel Alphayo Kambey
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Kouminin Kanwore
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Abiola Abdulrahman Ayanlaja
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Iqra Nadeem
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - YinZhen Du
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | | | - WenYa Liu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Dianshuai Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
17
|
Nguyen QL, Okuno N, Hamashima T, Dang ST, Fujikawa M, Ishii Y, Enomoto A, Maki T, Nguyen HN, Nguyen VT, Fujimori T, Mori H, Andrae J, Betsholtz C, Takao K, Yamamoto S, Sasahara M. Vascular PDGFR-alpha protects against BBB dysfunction after stroke in mice. Angiogenesis 2021; 24:35-46. [PMID: 32918673 DOI: 10.1007/s10456-020-09742-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023]
Abstract
Blood-brain barrier (BBB) dysfunction underlies the pathogenesis of many neurological diseases. Platelet-derived growth factor receptor-alpha (PDGFRα) induces hemorrhagic transformation (HT) downstream of tissue plasminogen activator in thrombolytic therapy of acute stroke. Thus, PDGFs are attractive therapeutic targets for BBB dysfunction. In the present study, we examined the role of PDGF signaling in the process of tissue remodeling after middle cerebral arterial occlusion (MCAO) in mice. Firstly, we found that imatinib increased lesion size after permanent MCAO in wild-type mice. Moreover, imatinib-induced HT only when administrated in the subacute phase of MCAO, but not in the acute phase. Secondly, we generated genetically mutated mice (C-KO mice) that showed decreased expression of perivascular PDGFRα. Additionally, transient MCAO experiments were performed in these mice. We found that the ischemic lesion size was not affected; however, the recruitment of PDGFRα/type I collagen-expressing perivascular cells was significantly downregulated, and HT and IgG leakage was augmented only in the subacute phase of stroke in C-KO mice. In both experiments, we found that the expression of tight junction proteins and PDGFRβ-expressing pericyte coverage was not significantly affected in imatinib-treated mice and in C-KO mice. The specific implication of PDGFRα signaling was suggestive of protective effects against BBB dysfunction during the subacute phase of stroke. Vascular TGF-β1 expression was downregulated in both imatinib-treated and C-KO mice, along with sustained levels of MMP9. Therefore, PDGFRα effects may be mediated by TGF-β1 which exerts potent protective effects in the BBB.
Collapse
Affiliation(s)
- Quang Linh Nguyen
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
- Stroke Center, The 108 Military Central Hospital, Ha Noi, Vietnam
| | - Noriko Okuno
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Takeru Hamashima
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Son Tung Dang
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Miwa Fujikawa
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Yoko Ishii
- Department of Health Science, Faculty of Health and Human Development, The University of Nagano, Nagano, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takakuni Maki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Van Tuyen Nguyen
- Stroke Center, The 108 Military Central Hospital, Ha Noi, Vietnam
| | - Toshihiko Fujimori
- Division of Embryology, National Institute for Basic Biology, Okazaki, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Johanna Andrae
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Integrated Cardio Metabolic Center, Karolinska Institute, Huddinge, Sweden
| | - Keizo Takao
- Division of Animal Resources and Development, Life Science Research Center, University of Toyama, Toyama, Japan
| | - Seiji Yamamoto
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.
| | - Masakiyo Sasahara
- Department of Pathology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.
| |
Collapse
|
18
|
Thompson A, Farmer K, Rowe E, Hayley S. Erythropoietin modulates striatal antioxidant signalling to reduce neurodegeneration in a toxicant model of Parkinson's disease. Mol Cell Neurosci 2020; 109:103554. [DOI: 10.1016/j.mcn.2020.103554] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
|
19
|
Sharma A, Muresanu DF, Castellani RJ, Nozari A, Lafuente JV, Sahib S, Tian ZR, Buzoianu AD, Patnaik R, Wiklund L, Sharma HS. Mild traumatic brain injury exacerbates Parkinson's disease induced hemeoxygenase-2 expression and brain pathology: Neuroprotective effects of co-administration of TiO 2 nanowired mesenchymal stem cells and cerebrolysin. PROGRESS IN BRAIN RESEARCH 2020; 258:157-231. [PMID: 33223035 DOI: 10.1016/bs.pbr.2020.09.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mild traumatic brain injury (mTBI) is one of the leading predisposing factors in the development of Parkinson's disease (PD). Mild or moderate TBI induces rapid production of tau protein and alpha synuclein (ASNC) in the cerebrospinal fluid (CSF) and in several brain areas. Enhanced tau-phosphorylation and ASNC alters the molecular machinery of the brain leading to PD pathology. Recent evidences show upregulation of constitutive isoform of hemeoxygenase (HO-2) in PD patients that correlates well with the brain pathology. mTBI alone induces profound upregulation of HO-2 immunoreactivity. Thus, it would be interesting to explore whether mTBI exacerbates PD pathology in relation to tau, ASNC and HO-2 expression. In addition, whether neurotrophic factors and stem cells known to reduce brain pathology in TBI could induce neuroprotection in PD following mTBI. In this review role of mesenchymal stem cells (MSCs) and cerebrolysin (CBL), a well-balanced composition of several neurotrophic factors and active peptide fragments using nanowired delivery in PD following mTBI is discussed based on our own investigation. Our results show that mTBI induces concussion exacerbates PD pathology and nanowired delivery of MSCs and CBL induces superior neuroprotection. This could be due to reduction in tau, ASNC and HO-2 expression in PD following mTBI, not reported earlier. The functional significance of our findings in relation to clinical strategies is discussed.
Collapse
Affiliation(s)
- Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
20
|
Richardson RM, Bankiewicz KS, Christine CW, Van Laar AD, Gross RE, Lonser R, Factor SA, Kostyk SK, Kells AP, Ravina B, Larson PS. Data-driven evolution of neurosurgical gene therapy delivery in Parkinson's disease. J Neurol Neurosurg Psychiatry 2020; 91:1210-1218. [PMID: 32732384 PMCID: PMC7569395 DOI: 10.1136/jnnp-2020-322904] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/28/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022]
Abstract
Loss of nigrostriatal dopaminergic projection neurons is a key pathology in Parkinson's disease, leading to abnormal function of basal ganglia motor circuits and the accompanying characteristic motor features. A number of intraparenchymally delivered gene therapies designed to modify underlying disease and/or improve clinical symptoms have shown promise in preclinical studies and subsequently were evaluated in clinical trials. Here we review the challenges with surgical delivery of gene therapy vectors that limited therapeutic outcomes in these trials, particularly the lack of real-time monitoring of vector administration. These challenges have recently been addressed during the evolution of novel techniques for vector delivery that include the use of intraoperative MRI. The preclinical development of these techniques are described in relation to recent clinical translation in an adeno-associated virus serotype 2-mediated human aromatic L-amino acid decarboxylase gene therapy development programme. This new paradigm allows visualisation of the accuracy and adequacy of viral vector delivery within target structures, enabling intertrial modifications in surgical approaches, cannula design, vector volumes and dosing. The rapid, data-driven evolution of these procedures is unique and has led to improved vector delivery.
Collapse
Affiliation(s)
- R Mark Richardson
- Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts, USA .,Harvard Medical School, Boston, Massachusetts, USA
| | - Krystof S Bankiewicz
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA.,Department of Neurological Surgery, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Chadwick W Christine
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Amber D Van Laar
- Department of Neurology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Brain Neurotherapy Bio, Inc, Columbus, Ohio, USA
| | - Robert E Gross
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA.,Department of Neurology, Emory University, Atlanta, Georgia, USA
| | - Russell Lonser
- Department of Neurological Surgery, Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Stewart A Factor
- Department of Neurology, Emory University, Atlanta, Georgia, USA
| | - Sandra K Kostyk
- Departments of Neuroscience and Neurology, Ohio State University College of Medicine, Columbus, Ohio, USA
| | | | - Bernard Ravina
- Praxis Precision Medicines, Inc, Cambridge, Massachusetts, USA
| | - Paul S Larson
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
21
|
Sidorova YA, Saarma M. Can Growth Factors Cure Parkinson's Disease? Trends Pharmacol Sci 2020; 41:909-922. [PMID: 33198924 DOI: 10.1016/j.tips.2020.09.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/25/2020] [Accepted: 09/30/2020] [Indexed: 01/03/2023]
Abstract
Growth factors (GFs) hold considerable promise for disease modification in neurodegenerative disorders because they can protect and restore degenerating neurons and also enhance their functional activity. However, extensive efforts applied to utilize their therapeutic potential in humans have achieved limited success so far. Multiple clinical trials with GFs were performed in Parkinson's disease (PD) patients, in whom diagnostic symptoms of the disease are caused by advanced degeneration of nigrostriatal dopamine neurons (DNs), but the results of these trials are controversial. This review discusses recent developments in the field of therapeutic use of GFs, problems and obstacles related to this use, suggests the ways to overcome these issues, and alternative approaches that can be used to utilize the potential ofGFsin PD management.
Collapse
Affiliation(s)
- Yulia A Sidorova
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
22
|
Congrong Shujing Granule-Induced GRP78 Expression Reduced Endoplasmic Reticulum Stress and Neuronal Apoptosis in the Midbrain in a Parkinson's Disease Rat Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:4796236. [PMID: 33062012 PMCID: PMC7547351 DOI: 10.1155/2020/4796236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/08/2020] [Accepted: 07/30/2020] [Indexed: 11/17/2022]
Abstract
The main pathological changes inherent in Parkinson's disease (PD) are degeneration and loss of dopamine neurons in the midbrain and formation of Lewy bodies. Many studies have shown that the pathogenesis of PD is closely related to endoplasmic reticulum (ER) oxidative stress. This study combined various traditional Chinese medicines to prepare Congrong Shujing granules (CSGs). The optimal dose combination of the ingredients was identified by experimental intervention in SH-SY5Y cells in vitro. A PD rat model was established by intraperitoneal injection of rotenone sunflower oil emulsion. The suspension tests were performed on the 14th day after modeling and also on the 14th day after CSG intervention (5.88 g/kg, 11.76 g/kg, and 23.52 g/kg). We evaluated the changes in motor function and the expression of neuronal cell functional marker proteins, ER stress (ERS) marker proteins, and apoptosis-related pathway proteins of neuronal cells. Changes in cellular ultrastructure were observed by electron microscopy. Our results showed that CSG treatment lengthened the duration of PD rats' gripping to the wire. 78 kDa glucose-regulated protein (GRP78) expression in the substantia nigra was significantly upregulated in the middle- and high-dose CSG groups after 14 days of treatment compared with the model group. The expression of the key dopaminergic neuron functional enzyme tyrosine hydroxylase (TH) and cerebral dopamine neurotrophic factor (CDNF) was elevated. The expression of c-Jun N-terminal kinase (JNK) and phosphorylated c-Jun decreased, and cell apoptosis was significantly reduced. Compared with the model group, the treatment groups had fewer ER fragmentation and degranulation (ribosome shedding) and abundant ER and mitochondria suggesting that CSG reduced ER stress and neuronal apoptosis in the midbrain of a PD rat model by inducing the expression of molecular chaperone GRP78.
Collapse
|
23
|
Anantha J, Goulding SR, Wyatt SL, Concannon RM, Collins LM, Sullivan AM, O'Keeffe GW. STRAP and NME1 Mediate the Neurite Growth-Promoting Effects of the Neurotrophic Factor GDF5. iScience 2020; 23:101457. [PMID: 32853992 PMCID: PMC7452236 DOI: 10.1016/j.isci.2020.101457] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/17/2020] [Accepted: 08/10/2020] [Indexed: 12/23/2022] Open
Abstract
Loss of midbrain dopaminergic (mDA) neurons and their axons is central to Parkinson's disease (PD). Growth differentiation factor (GDF)5 is a potential neurotrophic factor for PD therapy. However, the molecular mediators of its neurotrophic action are unknown. Our proteomics analysis shows that GDF5 increases the expression of serine threonine receptor-associated protein kinase (STRAP) and nucleoside diphosphate kinase (NME)1 in the SH-SY5Y neuronal cell line. GDF5 overexpression increased NME1 expression in adult rat brain in vivo. NME and STRAP mRNAs are expressed in developing and adult rodent midbrain. Expression of both STRAP and NME1 is necessary and sufficient for the promotion of neurite growth in SH-SY5Y cells by GDF5. NME1 treatment increased neurite growth in both SH-SY5Y cells and cultured mDA neurons. Expression patterns of NME and STRAP are altered in PD midbrain. NME1 and STRAP are thus key mediators of GDF5's neurotrophic effects, rationalizing their future study as therapeutic targets for PD.
Collapse
Affiliation(s)
- Jayanth Anantha
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
| | - Susan R. Goulding
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
- Department of Biological Sciences, Cork Institute of Technology, Cork, Ireland
| | - Sean L. Wyatt
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, UK
| | - Ruth M. Concannon
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
| | - Louise M. Collins
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
- Department of Physiology, UCC, Cork, Ireland
| | - Aideen M. Sullivan
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
- APC Microbiome Ireland, UCC, Cork, Ireland
- Cork Neuroscience Centre, UCC, Cork, Ireland
| | - Gerard W. O'Keeffe
- Department of Anatomy & Neuroscience, University College Cork (UCC), Cork, Ireland
- APC Microbiome Ireland, UCC, Cork, Ireland
- Cork Neuroscience Centre, UCC, Cork, Ireland
| |
Collapse
|
24
|
Leggio L, Paternò G, Vivarelli S, L’Episcopo F, Tirolo C, Raciti G, Pappalardo F, Giachino C, Caniglia S, Serapide MF, Marchetti B, Iraci N. Extracellular Vesicles as Nanotherapeutics for Parkinson's Disease. Biomolecules 2020; 10:E1327. [PMID: 32948090 PMCID: PMC7563168 DOI: 10.3390/biom10091327] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/10/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are naturally occurring membranous structures secreted by normal and diseased cells, and carrying a wide range of bioactive molecules. In the central nervous system (CNS), EVs are important in both homeostasis and pathology. Through receptor-ligand interactions, direct fusion, or endocytosis, EVs interact with their target cells. Accumulating evidence indicates that EVs play crucial roles in the pathogenesis of many neurodegenerative disorders (NDs), including Parkinson's disease (PD). PD is the second most common ND, characterized by the progressive loss of dopaminergic (DAergic) neurons within the Substantia Nigra pars compacta (SNpc). In PD, EVs are secreted by both neurons and glial cells, with either beneficial or detrimental effects, via a complex program of cell-to-cell communication. The functions of EVs in PD range from their etiopathogenetic relevance to their use as diagnostic tools and innovative carriers of therapeutics. Because they can cross the blood-brain barrier, EVs can be engineered to deliver bioactive molecules (e.g., small interfering RNAs, catalase) within the CNS. This review summarizes the latest findings regarding the role played by EVs in PD etiology, diagnosis, prognosis, and therapy, with a particular focus on their use as novel PD nanotherapeutics.
Collapse
Affiliation(s)
- Loredana Leggio
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (L.L.); (G.P.); (S.V.); (G.R.); (F.P.); (M.F.S.)
| | - Greta Paternò
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (L.L.); (G.P.); (S.V.); (G.R.); (F.P.); (M.F.S.)
| | - Silvia Vivarelli
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (L.L.); (G.P.); (S.V.); (G.R.); (F.P.); (M.F.S.)
| | - Francesca L’Episcopo
- Neuropharmacology Section, OASI Research Institute-IRCCS, 94018 Troina, Italy; (F.L.); (C.T.); (C.G.); (S.C.)
| | - Cataldo Tirolo
- Neuropharmacology Section, OASI Research Institute-IRCCS, 94018 Troina, Italy; (F.L.); (C.T.); (C.G.); (S.C.)
| | - Gabriele Raciti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (L.L.); (G.P.); (S.V.); (G.R.); (F.P.); (M.F.S.)
| | - Fabrizio Pappalardo
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (L.L.); (G.P.); (S.V.); (G.R.); (F.P.); (M.F.S.)
| | - Carmela Giachino
- Neuropharmacology Section, OASI Research Institute-IRCCS, 94018 Troina, Italy; (F.L.); (C.T.); (C.G.); (S.C.)
| | - Salvatore Caniglia
- Neuropharmacology Section, OASI Research Institute-IRCCS, 94018 Troina, Italy; (F.L.); (C.T.); (C.G.); (S.C.)
| | - Maria Francesca Serapide
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (L.L.); (G.P.); (S.V.); (G.R.); (F.P.); (M.F.S.)
| | - Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (L.L.); (G.P.); (S.V.); (G.R.); (F.P.); (M.F.S.)
- Neuropharmacology Section, OASI Research Institute-IRCCS, 94018 Troina, Italy; (F.L.); (C.T.); (C.G.); (S.C.)
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Torre Biologica, Via Santa Sofia 97, 95125 Catania, Italy; (L.L.); (G.P.); (S.V.); (G.R.); (F.P.); (M.F.S.)
| |
Collapse
|
25
|
Lombaert IMA, Patel VN, Jones CE, Villier DC, Canada AE, Moore MR, Berenstein E, Zheng C, Goldsmith CM, Chorini JA, Martin D, Zourelias L, Trombetta MG, Edwards PC, Meyer K, Ando D, Passineau MJ, Hoffman MP. CERE-120 Prevents Irradiation-Induced Hypofunction and Restores Immune Homeostasis in Porcine Salivary Glands. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:839-855. [PMID: 32953934 PMCID: PMC7479444 DOI: 10.1016/j.omtm.2020.07.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023]
Abstract
Salivary gland hypofunction causes significant morbidity and loss of quality of life for head and neck cancer patients treated with radiotherapy. Preventing hypofunction is an unmet therapeutic need. We used an adeno-associated virus serotype 2 (AAV2) vector expressing the human neurotrophic factor neurturin (CERE-120) to treat murine submandibular glands either pre- or post-irradiation (IR). Treatment with CERE-120 pre-IR, not post-IR, prevented hypofunction. RNA sequencing (RNA-seq) analysis showed reduced gene expression associated with fibrosis and the innate and humoral immune responses. We then used a minipig model with CERE-120 treatment pre-IR and also compared outcomes of the contralateral non-IR gland. Analysis of gene expression, morphology, and immunostaining showed reduced IR-related immune responses and improved secretory mechanisms. CERE-120 prevented IR-induced hypofunction and restored immune homeostasis, and there was a coordinated contralateral gland response to either damage or treatment. CERE-120 gene therapy is a potential treatment for head and neck cancer patients to influence communication among neuronal, immune, and epithelial cells to prevent IR-induced salivary hypofunction and restore immune homeostasis.
Collapse
Affiliation(s)
- Isabelle M A Lombaert
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD 20892, USA.,Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vaishali N Patel
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD 20892, USA
| | - Christina E Jones
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD 20892, USA
| | - Derrick C Villier
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD 20892, USA
| | - Ashley E Canada
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD 20892, USA
| | - Matthew R Moore
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD 20892, USA
| | - Elsa Berenstein
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD 20892, USA
| | - Changyu Zheng
- Translational Research Core, NIDCR, NIH, DHHS, Bethesda, MD 20892, USA
| | | | - John A Chorini
- Adeno-Associated Virus Section, NIDCR, NIH, DHHS, Bethesda, MD 20892, USA
| | - Daniel Martin
- Genomics and Computational Biology Core, NIDCR, NIH, DHHS, Bethesda, MD 20892, USA
| | - Lee Zourelias
- Gene Therapy Program, Department of Medicine, Division of Cardiovascular Medicine, Allegheny Health Network, Pittsburg, PA 15212, USA
| | - Mark G Trombetta
- Department of Oncology, Division of Radiation Oncology, Allegheny Health Network, Pittsburg, PA 15212, USA
| | - Paul C Edwards
- Department of Oral Pathology, Medicine, and Radiology, Indiana University School of Dentistry, Indianapolis, IN 46202, USA
| | - Kathleen Meyer
- Sangamo BioSciences, Inc., 501 Canal Blvd., Richmond, CA 94804
| | - Dale Ando
- Sangamo BioSciences, Inc., 501 Canal Blvd., Richmond, CA 94804
| | - Michael J Passineau
- Gene Therapy Program, Department of Medicine, Division of Cardiovascular Medicine, Allegheny Health Network, Pittsburg, PA 15212, USA
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, NIH, DHHS, Bethesda, MD 20892, USA
| |
Collapse
|
26
|
Chmielarz P, Saarma M. Neurotrophic factors for disease-modifying treatments of Parkinson's disease: gaps between basic science and clinical studies. Pharmacol Rep 2020; 72:1195-1217. [PMID: 32700249 PMCID: PMC7550372 DOI: 10.1007/s43440-020-00120-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 06/16/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023]
Abstract
Abstract Background Neurotrophic factors are endogenous proteins promoting the survival of different neural cells. Therefore, they elicited great interest as a possible treatment for neurodegenerative disorders, including Parkinson’s Disease (PD). PD is the second most common neurodegenerative disorder, scientifically characterized more than 200 years ago and initially linked with motor abnormalities. Currently, the disease is viewed as a highly heterogeneous, progressive disorder with a long presymptomatic phase, and both motor and non-motor symptoms. Presently only symptomatic treatments for PD are available. Neurohistopathological changes of PD affected brains have been described more than 100 years ago and characterized by the presence of proteinaceous inclusions known as Lewy bodies and degeneration of dopamine neurons. Despite more than a century of investigations, it has remained unclear why dopamine neurons die in PD. Methods This review summarizes literature data from preclinical studies and clinical trials of neurotrophic factor based therapies for PD and discuss it from the perspective of the current understanding of PD biology. Results Newest data point towards dysfunctions of mitochondria, autophagy-lysosomal pathway, unfolded protein response and prion protein-like spreading of misfolded alpha-synuclein that is the major component of Lewy bodies. Yet, the exact chain of events leading to the demise of dopamine neurons is unclear and perhaps different in subpopulations of patients. Conclusions Gaps in our understanding of underlying disease etiology have hindered our attempts to find treatments able to slow down the progression of PD. Graphic abstract ![]()
Collapse
Affiliation(s)
- Piotr Chmielarz
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
27
|
Zoumboulakis D, Cirella KR, Gougeon PY, Lourenssen SR, Blennerhassett MG. MMP-9 Processing of Intestinal Smooth Muscle-derived GDNF is Required for Neurotrophic Action on Enteric Neurons. Neuroscience 2020; 443:8-18. [PMID: 32682824 DOI: 10.1016/j.neuroscience.2020.07.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/09/2020] [Accepted: 07/11/2020] [Indexed: 12/11/2022]
Abstract
The neurotrophin GDNF guides development of the enteric nervous system (ENS) in embryogenesis and directs survival and axon outgrowth in postnatal myenteric neurons in vitro. GDNF expression in intestinal smooth muscle cells is dynamic, with upregulation by inflammatory cytokines in vitro or intestinal inflammation in vivo, but the role of post-translational proteolytic cleavage is undefined. In a co-culture model of myenteric neurons, smooth muscle and glia, inhibition of serine or cysteine protease activity was ineffective against the >2-fold increase in axon density caused by TNFα. However, inhibitors of metalloproteinases (MMP) identified an essential role of MMP-9, and qPCR and western blotting showed that pro-inflammatory cytokines increased both mRNA and protein expression for MMP-9, in both cellular lysates and conditioned medium (CM). Inhibition of MMP-9 prevented the cytokine-induced increase in mature GDNF in CM or cellular lysates of co-cultures or cell lines of intestinal smooth muscle cells (ISMC) from adult rat colon. Western blotting showed parallel upregulation of mature GDNF and MMP-9 vs control in ISMC isolated on Day 2 of TNBS-induced colitis. Nonetheless, transfection of GDNF plasmid into HEK-293 cells as a carrier system, or directly into the co-culture model, conveyed a strong neurotrophic effect that was MMP-9 dependent. We conclude that MMP-9 activity is required for the neurotrophic effects of GDNF on myenteric neurons in vitro. However, the coordinated upregulation of GDNF and MMP-9 in intestinal smooth muscle by inflammatory cytokines provides a supportive, target cell-derived environment that limits inflammatory damage to the ENS.
Collapse
Affiliation(s)
- Demetri Zoumboulakis
- Gastrointestinal Diseases Research Unit and Queen's University, Kingston, ON K7L 2V7, Canada
| | - Kirsten R Cirella
- Gastrointestinal Diseases Research Unit and Queen's University, Kingston, ON K7L 2V7, Canada
| | - Pierre-Yves Gougeon
- Gastrointestinal Diseases Research Unit and Queen's University, Kingston, ON K7L 2V7, Canada
| | - Sandra R Lourenssen
- Gastrointestinal Diseases Research Unit and Queen's University, Kingston, ON K7L 2V7, Canada
| | | |
Collapse
|
28
|
An In Vitro Partial Lesion Model of Differentiated Human Mesencephalic Neurons: Effect of Pericyte Secretome on Phenotypic Markers. J Mol Neurosci 2020; 70:1914-1925. [PMID: 32472394 PMCID: PMC7561585 DOI: 10.1007/s12031-020-01589-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/13/2020] [Indexed: 11/17/2022]
Abstract
Parkinson’s disease (PD) is characterised by the progressive degeneration of dopaminergic (DA) neurons in the substantia nigra pars compacta. Post-mortem data suggests that the loss of DA markers may long precede the cell death, leaving a window to rescue the DA phenotype. Screening for potential neuroprotective or restorative therapies, however, requires that partial lesions of DA neurons can be modelled in vitro. In order to establish a partial lesion model of DA neurons in vitro, we evaluated the effects of different exposure times to 1-methyl-4-phenylpyridinium (MPP+) and 6-hydroxydopamine (6-OHDA) on the cell survival and DA marker expression using DA neurons derived from the Lund human mesencephalic (LUHMES) cell line. We show that 24-h incubation with 50 μM of MPP+ or 6-h incubation with 100 μM of 6-OHDA leads to a significant decrease in the protein expression of DA markers without affecting overall cell death, consistent with a mild DA lesion. Using conditioned medium of human brain–derived pericytes stimulated with platelet-derived growth factor BB (PDGF-BB), we demonstrate a significant upregulation of DA markers. In conclusion, we provide an experimental model of an in vitro DA neuron partial lesion suitable to study different molecules and their potential neuroprotective or neurorestorative effects on the DA phenotype. We provide evidence that the secretome of brain pericytes stimulated via PDGF-BB/PDGFRβ affects DA marker expression and may represent one possible mechanism contributing to the neurorestoration previously observed in PD by this growth factor.
Collapse
|
29
|
Benoit SM, Xu H, Schmid S, Alexandrova R, Kaur G, Thiruvahindrapuram B, Pereira SL, Jog M, Hebb MO. Expanding the search for genetic biomarkers of Parkinson's disease into the living brain. Neurobiol Dis 2020; 140:104872. [PMID: 32302674 DOI: 10.1016/j.nbd.2020.104872] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 12/12/2022] Open
Abstract
Altered gene expression related to Parkinson's Disease (PD) has not been described in the living brain, yet this information may support novel discovery pertinent to disease pathophysiology and treatment. This study compared the transcriptome in brain biopsies obtained from living PD and Control patients. To evaluate the novelty of this data, a comprehensive literature review also compared differentially expressed gene (DEGs) identified in the current study with those reported in PD cadaveric brain and peripheral tissues. RNA was extracted from rapidly cryopreserved frontal lobe specimens collected from PD and Control patients undergoing neurosurgical procedures. RNA sequencing (RNA-Seq) was performed and validated using quantitative polymerase chain reaction. DEG data was assessed using bioinformatics and subsequently included within a comparative analysis of PD RNA-Seq studies. 370 DEGs identified in living brain specimens reflected diverse gene groups and included key members of trophic signaling, apoptosis, inflammation and cell metabolism pathways. The comprehensive literature review yielded 7 RNA-Seq datasets generated from blood, skin and cadaveric brain but none from a living brain source. From the current dataset, 123 DEGs were identified only within the living brain and 267 DEGs were either newly found or had distinct directional change in living brain relative to other tissues. This is the first known study to analyze the transcriptome in brain tissue from living PD and Control patients. The data produced using these methods offer a unique, unexplored resource with potential to advance insight into the genetic associations of PD.
Collapse
Affiliation(s)
- Simon M Benoit
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, 339 Windermere Road, Suite C7-134, London N6A 5A5, Ontario, Canada
| | - Hu Xu
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, 339 Windermere Road, Suite C7-134, London N6A 5A5, Ontario, Canada
| | - Susanne Schmid
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, Western University, 1151 Richmond Street, Medical Sciences Building, Room 443, London N6A 3K7, Ontario, Canada
| | - Roumiana Alexandrova
- The Centre for Applied Genomics, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 656 Bay Street, Room 139800, Toronto M5G 0A4, Ontario, Canada
| | - Gaganjot Kaur
- The Centre for Applied Genomics, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 656 Bay Street, Room 139800, Toronto M5G 0A4, Ontario, Canada
| | - Bhooma Thiruvahindrapuram
- The Centre for Applied Genomics, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 656 Bay Street, Room 139800, Toronto M5G 0A4, Ontario, Canada
| | - Sergio L Pereira
- The Centre for Applied Genomics, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 656 Bay Street, Room 139800, Toronto M5G 0A4, Ontario, Canada
| | - Mandar Jog
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, 339 Windermere Road, Suite C7-134, London N6A 5A5, Ontario, Canada
| | - Matthew O Hebb
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, 339 Windermere Road, Suite C7-134, London N6A 5A5, Ontario, Canada; Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, Western University, 1151 Richmond Street, Medical Sciences Building, Room 443, London N6A 3K7, Ontario, Canada.
| |
Collapse
|
30
|
Barker RA, Björklund A, Gash DM, Whone A, Van Laar A, Kordower JH, Bankiewicz K, Kieburtz K, Saarma M, Booms S, Huttunen HJ, Kells AP, Fiandaca MS, Stoessl AJ, Eidelberg D, Federoff H, Voutilainen MH, Dexter DT, Eberling J, Brundin P, Isaacs L, Mursaleen L, Bresolin E, Carroll C, Coles A, Fiske B, Matthews H, Lungu C, Wyse RK, Stott S, Lang AE. GDNF and Parkinson's Disease: Where Next? A Summary from a Recent Workshop. JOURNAL OF PARKINSON'S DISEASE 2020; 10:875-891. [PMID: 32508331 PMCID: PMC7458523 DOI: 10.3233/jpd-202004] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/18/2020] [Indexed: 12/22/2022]
Abstract
The concept of repairing the brain with growth factors has been pursued for many years in a variety of neurodegenerative diseases including primarily Parkinson's disease (PD) using glial cell line-derived neurotrophic factor (GDNF). This neurotrophic factor was discovered in 1993 and shown to have selective effects on promoting survival and regeneration of certain populations of neurons including the dopaminergic nigrostriatal pathway. These observations led to a series of clinical trials in PD patients including using infusions or gene delivery of GDNF or the related growth factor, neurturin (NRTN). Initial studies, some of which were open label, suggested that this approach could be of value in PD when the agent was injected into the putamen rather than the cerebral ventricles. In subsequent double-blind, placebo-controlled trials, the most recent reporting in 2019, treatment with GDNF did not achieve its primary end point. As a result, there has been uncertainty as to whether GDNF (and by extrapolation, related GDNF family neurotrophic factors) has merit in the future treatment of PD. To critically appraise the existing work and its future, a special workshop was held to discuss and debate this issue. This paper is a summary of that meeting with recommendations on whether there is a future for this therapeutic approach and also what any future PD trial involving GDNF and other GDNF family neurotrophic factors should consider in its design.
Collapse
Affiliation(s)
- Roger A. Barker
- Cambridge Centre for Brain Repair, Department of Clinical Neuroscience and WT-MRC Cambridge Stem Cell Institute, Cambridge, UK
| | | | - Don M. Gash
- Professor Emeritus of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Alan Whone
- Translational Health Sciences, Bristol Medical School, University of Bristol and Neurological and Musculoskeletal Sciences Division, North Bristol NHS Trust, Bristol, UK
| | | | - Jeffrey H. Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Krystof Bankiewicz
- Neurological Surgery, Gilbert and Kathryn Mitchell Endowed Chair, Director, Brain Health and Performance Center, The Ohio State University, Department of Neurological Surgery, Columbus, OH, USA
| | - Karl Kieburtz
- Center for Health & Technology, and the Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | - Henri J. Huttunen
- Herantis Pharma Plc, Finland
- Neuroscience Center, HiLIFE, University of Helsinki, Finland
| | | | | | - A. Jon Stoessl
- Pacific Parkinson’s Research Centre & Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Howard Federoff
- School of Medicine, Susan and Henry College of Health Sciences, University of California, Irvine and CEO, Aspen Neuroscience, San Diego, CA, USA
| | | | | | - Jamie Eberling
- The Michael J. Fox Foundation for Parkinson’s Research, New York, NY, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | | | - Leah Mursaleen
- The Cure Parkinson’s Trust, London, UK
- School of Life Sciences, University of Westminster, UK and School of Pharmacy, University College London, UK
| | | | | | - Alasdair Coles
- Department of Clinical Neuroscience, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| | - Brian Fiske
- The Michael J. Fox Foundation for Parkinson’s Research, New York, NY, USA
| | | | - Codrin Lungu
- Division of Clinical Research, National Institute of Neurological Disorders and Stroke, Rockville, MD, USA
| | | | | | - Anthony E. Lang
- The Edmond J Safra Program in Parkinson’s Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, and the Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Abstract
Parkinson's disease (PD) is an aging-related neurodegenerative disorder characterized by progressive motor impairment.The etiology of PD is poorly understood but likely involves both genetic and environmental factors; the management of the disease is still with symptomatic therapy without any interference on the progression of neurodegeneration. In the past two decades, the results of a series of prospective cohort studies suggested that lifestyle factors likely modify the risk of developing PD. Among these, physical activity is known to reduce the risk of a wide range of diseases and conditions, including cardiovascular disease, stroke, and diabetes.Recently, a growing body of evidence has suggested that increased physical activity may also reduce the risk of PD and partly improve motor and non-motor symptoms during the disease course.Here we report the main findings on the effect of physical activity on both mobility and cognition either in animal models of PD or in people with PD. We also highlighted the structural and functional links between gait and cognition by reporting evidence from neuroimaging studies.
Collapse
Affiliation(s)
- Simona Bonavita
- II Clinic of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
32
|
Chen C, Guderyon MJ, Li Y, Ge G, Bhattacharjee A, Ballard C, He Z, Masliah E, Clark RA, O'Connor JC, Li S. Non-toxic HSC Transplantation-Based Macrophage/Microglia-Mediated GDNF Delivery for Parkinson's Disease. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 17:83-98. [PMID: 31890743 PMCID: PMC6931095 DOI: 10.1016/j.omtm.2019.11.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/15/2019] [Indexed: 02/08/2023]
Abstract
Glial cell-line-derived neurotrophic factor (GDNF) is a potent neuroprotective agent in cellular and animal models of Parkinson’s disease (PD). However, CNS delivery of GDNF in clinical trials has proven challenging due to blood-brain barrier (BBB) impermeability, poor diffusion within brain tissue, and large brain size. We report that using non-toxic mobilization-enabled preconditioning, hematopoietic stem cell (HSC) transplantation-based macrophage-mediated gene delivery may provide a solution to overcome these obstacles. Syngeneic bone marrow HSCs were transduced ex vivo with a lentiviral vector expressing macrophage promoter-driven GDNF and transplanted into 14-week-old MitoPark mice exhibiting PD-like impairments. Transplant preconditioning with granulocyte colony-stimulating factor (G-CSF) and AMD3100 was used to vacate bone marrow stem cell niches. Chimerism reached ∼80% after seven transplantation cycles. Transgene-expressing macrophages infiltrated degenerating CNS regions of MitoPark mice (not wild-type littermate controls), resulting in increased GDNF levels in the midbrain. Macrophage GDNF delivery not only markedly improved motor and non-motor dysfunction, but also dramatically mitigated the loss of dopaminergic neurons in both substantia nigra and the ventral tegmental area and preserved axonal terminals in the striatum. Striatal dopamine levels were almost completely restored. Our data support further development of mobilization-enabled HSC transplantation (HSCT)-based macrophage-mediated GDNF gene delivery as a disease-modifying therapy for PD.
Collapse
Affiliation(s)
- Cang Chen
- Department of Medicine, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Michael J Guderyon
- Department of Medicine, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Yang Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo Ge
- Department of Medicine, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Anindita Bhattacharjee
- Department of Medicine, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Cori Ballard
- Department of Medicine, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Zhixu He
- Department of Pediatrics, Zunyi Medical University Affiliated Hospital and Key Laboratory of Adult Stem Cell Transformation Research, Chinese Academy of Medical Science, Guiyang, Guizhou 550025, China
| | | | - Robert A Clark
- Department of Medicine, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.,Audie L. Murphy VA Hospital, 7400 Merton Minter Boulevard, San Antonio, TX 78229, USA
| | - Jason C O'Connor
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.,Audie L. Murphy VA Hospital, 7400 Merton Minter Boulevard, San Antonio, TX 78229, USA
| | - Senlin Li
- Department of Medicine, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.,Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.,Audie L. Murphy VA Hospital, 7400 Merton Minter Boulevard, San Antonio, TX 78229, USA
| |
Collapse
|
33
|
Robinson EJ, Aguiar S, Smidt MP, van der Heide LP. MCL1 as a Therapeutic Target in Parkinson's Disease? Trends Mol Med 2019; 25:1056-1065. [PMID: 31706839 DOI: 10.1016/j.molmed.2019.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/29/2019] [Accepted: 08/27/2019] [Indexed: 12/26/2022]
Abstract
Dopamine neurons in the substantia nigra (SN) pars compacta are selectively lost during the progression of Parkinson's disease (PD). Recent work performed on the role of the Bcl2 family (highly specialized proteins which control cellular survival and death) in midbrain dopamine neurons has led to the identification of the Bcl2 factor Mcl1 as a weak link in the survival of these neurons. We hypothesize that the regulation of BCL2 proteins may explain this selective vulnerability, and may even provide a novel therapeutic opportunity - strengthening weak links such as MCL1 could result in a delay or complete abrogation of cell death during PD.
Collapse
Affiliation(s)
- Edward J Robinson
- Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands
| | - Sebastian Aguiar
- Ageing and Cellular Senescence Laboratory, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, UK
| | - Marten P Smidt
- Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands
| | - Lars P van der Heide
- Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, the Netherlands.
| |
Collapse
|
34
|
Emir GK, Ünal Y, Yılmaz N, Tosun K, Kutlu G. The association of low levels of nesfatin-1 and glucagon-like peptide-1 with oxidative stress in Parkinson’s disease. Neurol Sci 2019; 40:2529-2535. [PMID: 31280388 DOI: 10.1007/s10072-019-03975-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/07/2019] [Indexed: 12/14/2022]
|
35
|
Abstract
Neurotrophic factors (NTF) are a subgroup of growth factors that promote survival and
differentiation of neurons. Due to their neuroprotective and neurorestorative properties,
their therapeutic potential has been tested in various neurodegenerative diseases.
Bioavailability of NTFs in the target tissue remains a major challenge for NTF-based
therapies. Various intracerebral delivery approaches, both protein and gene
transfer-based, have been tested with varying outcomes. Three growth factors, glial
cell-line derived neurotrophic factor (GDNF), neurturin (NRTN) and platelet-derived growth
factor (PDGF-BB) have been tested in clinical trials in Parkinson’s disease (PD) during
the past 20 years. A new protein can now be added to this list, as cerebral dopamine
neurotrophic factor (CDNF) has recently entered clinical trials. Despite their misleading
names, CDNF, together with its closest relative mesencephalic astrocyte-derived
neurotrophic factor (MANF), form a novel family of unconventional NTF that are both
structurally and mechanistically distinct from other growth factors. CDNF and MANF are
localized mainly to the lumen of endoplasmic reticulum (ER) and their primary function
appears to be modulation of the unfolded protein response (UPR) pathway. Prolonged ER
stress, via the UPR signaling pathways, contributes to the pathogenesis in a number of
chronic degenerative diseases, and is an important target for therapeutic modulation.
Intraputamenally administered recombinant human CDNF has shown robust neurorestorative
effects in a number of small and large animal models of PD, and had a good safety profile
in preclinical toxicology studies. Intermittent monthly bilateral intraputamenal infusions
of CDNF are currently being tested in a randomized placebo-controlled phase I–II clinical
study in moderately advanced PD patients. Here, we review the history of growth
factor-based clinical trials in PD, and discuss how CDNF differs from the previously
tested growth factors.
Collapse
Affiliation(s)
- Henri J Huttunen
- 1 Herantis Pharma Plc, Espoo, Finland.,2 Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- 3 Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
36
|
Nanodelivery of cerebrolysin reduces pathophysiology of Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2019; 245:201-246. [DOI: 10.1016/bs.pbr.2019.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
37
|
Crowley EK, Nolan YM, Sullivan AM. Exercise as a therapeutic intervention for motor and non-motor symptoms in Parkinson's disease: Evidence from rodent models. Prog Neurobiol 2018; 172:2-22. [PMID: 30481560 DOI: 10.1016/j.pneurobio.2018.11.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 10/25/2018] [Accepted: 11/23/2018] [Indexed: 12/11/2022]
Abstract
Parkinson's disease (PD) is characterised by degeneration of dopaminergic neurons of the nigrostriatal pathway, which leads to the cardinal motor symptoms of the disease - tremor, rigidity and postural instability. A number of non-motor symptoms are also associated with PD, including cognitive impairment, mood disturbances and dysfunction of gastrointestinal and autonomic systems. Current therapies provide symptomatic relief but do not halt the disease process, so there is an urgent need for preventative strategies. Lifestyle interventions such as aerobic exercise have shown potential to lower the risk of developing PD and to alleviate both motor and non-motor symptoms. However, there is a lack of large-scale randomised clinical trials that have employed exercise in PD patients. This review will focus on the evidence from studies on rodent models of PD, for employing exercise as an intervention for both motor and non-motor symptoms.
Collapse
Affiliation(s)
- E K Crowley
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Y M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland
| | - A M Sullivan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland.
| |
Collapse
|